1
|
The sustained PGE 2 release matrix improves neovascularization and skeletal muscle regeneration in a hindlimb ischemia model. J Nanobiotechnology 2022; 20:95. [PMID: 35209908 PMCID: PMC8867652 DOI: 10.1186/s12951-022-01301-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 02/06/2022] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND The promising therapeutic strategy for the treatment of peripheral artery disease (PAD) is to restore blood supply and promote regeneration of skeletal muscle regeneration. Increasing evidence revealed that prostaglandin E2 (PGE2), a lipid signaling molecule, has significant therapeutic potential for tissue repair and regeneration. Though PGE2 has been well reported in tissue regeneration, the application of PGE2 is hampered by its short half-life in vivo and the lack of a viable system for sustained release of PGE2. RESULTS In this study, we designed and synthesized a new PGE2 release matrix by chemically bonding PGE2 to collagen. Our results revealed that the PGE2 matrix effectively extends the half-life of PGE2 in vitro and in vivo. Moreover, the PGE2 matrix markedly improved neovascularization by increasing angiogenesis, as confirmed by bioluminescence imaging (BLI). Furthermore, the PGE2 matrix exhibits superior therapeutic efficacy in the hindlimb ischemia model through the activation of MyoD1-mediated muscle stem cells, which is consistent with accelerated structural recovery of skeletal muscle, as evidenced by histological analysis. CONCLUSIONS Our findings highlight the chemical bonding strategy of chemical bonding PGE2 to collagen for sustained release and may facilitate the development of PGE2-based therapies to significantly improve tissue regeneration.
Collapse
|
2
|
Liu Y, Huang Z, Li Z. Molecular Imaging of Tumor Progression and Angiogenesis by Dual Bioluminescence. Methods Mol Biol 2022; 2524:457-469. [PMID: 35821492 DOI: 10.1007/978-1-0716-2453-1_34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Angiogenesis is a prerequisite for tumor growth and invasion, and anti-angiogenesis has become a highlight in tumor treatment research. However, so far, there is no reliable solution for how to simultaneously visualize the relationship between tumor progression and angiogenesis. Bioluminescence imaging (BLI) has been broadly utilized and is a very promising non-invasive imaging technique with the advantages of low cost, high sensitivity, and robust specificity. In this chapter, we describe a dual bioluminescence imaging BLI protocol for tumor progression and angiogenesis through implanting murine breast cancer cell line 4T1 which stably expressing Renilla luciferase (RLuc) into the transgenic mice with angiogenesis-induced firefly luciferase (FLuc) expression. This modality enables us to synchronously monitor the tumor progression and angiogenesis in the same mouse, which has broad applicability in oncology studies.
Collapse
Affiliation(s)
- Yue Liu
- School of Medicine, Nankai University, Tianjin, China
- The Key Laboratory of Bioactive Materials, The College of Life Science, Ministry of Education, Nankai University, Tianjin, China
| | - Ziyu Huang
- School of Medicine, Nankai University, Tianjin, China
- The Key Laboratory of Bioactive Materials, The College of Life Science, Ministry of Education, Nankai University, Tianjin, China
| | - Zongjin Li
- School of Medicine, Nankai University, Tianjin, China.
- The Key Laboratory of Bioactive Materials, The College of Life Science, Ministry of Education, Nankai University, Tianjin, China.
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Nankai University Affiliated Hospital of Obstetrics and Gynecology, Tianjin, China.
- State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
3
|
Alsawaftah N, Farooq A, Dhou S, Majdalawieh AF. Bioluminescence Imaging Applications in Cancer: A Comprehensive Review. IEEE Rev Biomed Eng 2021; 14:307-326. [PMID: 32746363 DOI: 10.1109/rbme.2020.2995124] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
Bioluminescence imaging (BLI), an optical preclinical imaging modality, is an invaluable imaging modality due to its low-cost, high throughput, fast acquisition times, and functional imaging capabilities. BLI is being extensively used in the field of cancer imaging, especially with the recent developments in genetic-engineering, stem cell, and gene therapy treatments. The purpose of this paper is to provide a comprehensive review of the principles, developments, and current status of BLI in cancer research. This paper covers the fundamental BLI concepts including BLI reporters and enzyme-substrate systems, data acquisition, and image characteristics. It reviews the studies discussing the use of BLI in cancer research such as imaging tumor-characteristic phenomena including tumorigenesis, metastasis, cancer metabolism, apoptosis, hypoxia, and angiogenesis, and response to cancer therapy treatments including chemotherapy, radiotherapy, immunotherapy, gene therapy, and stem cell therapy. The key advantages and disadvantages of BLI compared to other common imaging modalities are also discussed.
Collapse
|
4
|
Zhang K, Chen X, Li H, Feng G, Nie Y, Wei Y, Li N, Han Z, Han ZC, Kong D, Guo Z, Zhao Q, Li Z. A nitric oxide-releasing hydrogel for enhancing the therapeutic effects of mesenchymal stem cell therapy for hindlimb ischemia. Acta Biomater 2020; 113:289-304. [PMID: 32663662 DOI: 10.1016/j.actbio.2020.07.011] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 07/04/2020] [Accepted: 07/07/2020] [Indexed: 02/06/2023]
Abstract
Therapeutic angiogenesis with mesenchymal stem cells (MSCs) is promising for the clinical treatment of peripheral artery disease (PAD). However, the heterogeneous proangiogenic nature of MSCs is a key challenge in developing more effective treatments with MSCs for therapeutic angiogenesis purposes. Here, we propose to enhance the therapeutic function of human placenta-derived MSCs (hP-MSCs) in hindlimb ischemia therapy by using nitric oxide (NO)-releasing chitosan hydrogel (CS-NO). Our data showed that the co-transplantation of CS-NO hydrogel with hP-MSCs remarkably improved the grafting of hP-MSCs and ameliorated the functional recovery of ischemic hindlimbs. Moreover, we found that the neovascularization of damaged hindlimbs was significantly increased after co-transplanting CS-NO hydrogel and hP-MSCs, as confirmed by bioluminescence imaging (BLI). Further analysis revealed an endothelial-like status transformation of hP-MSCs in the presence of NO, which was identified as a potential mechanism contributing to the enhanced endothelium-protective and proangiogenic capacities of hP-MSCs that promote angiogenesis in mouse models of hindlimb ischemia. In conclusion, this study provides a promising approach for using NO hydrogel to improve the proangiogenic potency of MSCs in ischemic diseases, and the strategy used here facilitates the development of controlled-release scaffolds for enhancing the therapeutic efficiency of MSCs in angiogenic therapy. STATEMENT OF SIGNIFICANCE: The heterogeneous proangiogenic nature of mesenchymal stem cells (MSCs) is a key challenge in developing more effective treatments with MSCs for therapeutic angiogenesis purposes. In this study, we investigated whether nitric oxide (NO)-releasing chitosan hydrogel (CS-NO) could improve the proangiogenic potency of MSCs in ischemic diseases. Our results revealed an endothelial-like status transformation of human placenta-derived MSCs (hP-MSCs) in the presence of NO, which was identified as a potential mechanism contributing to the enhanced endothelium-protective and proangiogenic capacities of hP-MSCs that promote angiogenesis in mouse models of hindlimb ischemia. The strategy for enhancing the pro-angiogenic activity of MSCs with biomaterials provides a practical idea for overcoming the challenges associated with the clinical application of MSCs in therapeutic angiogenesis.
Collapse
Affiliation(s)
- Kaiyue Zhang
- Nankai University School of Medicine, 94 Weijin Road, Tianjin, China; State Key Laboratory of Medicinal Chemical Biology, The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, the College of Life Sciences, 94 Weijin Road, Tianjin 300071, China
| | - Xiaoniao Chen
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China; State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, China
| | - Huifang Li
- Nankai University School of Medicine, 94 Weijin Road, Tianjin, China
| | - Guowei Feng
- Nankai University School of Medicine, 94 Weijin Road, Tianjin, China
| | - Yan Nie
- Nankai University School of Medicine, 94 Weijin Road, Tianjin, China
| | - Yongzhen Wei
- State Key Laboratory of Medicinal Chemical Biology, The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, the College of Life Sciences, 94 Weijin Road, Tianjin 300071, China
| | - Nana Li
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, 601 Jinsui Road, Xinxiang, Henan 453003, China
| | - Zhibo Han
- Jiangxi Engineering Research Center for Stem Cell, Shangrao, Jiangxi, China; Tianjin Key Laboratory of Engineering Technologies for Cell Pharmaceutical, National Engineering Research Center of Cell Products, AmCellGene Co., Ltd., Tianjin, China; Beijing Engineering Laboratory of Perinatal Stem Cells, Beijing Institute of Health and Stem Cells, Health & Biotech Co., Beijing, China
| | - Zhong-Chao Han
- Jiangxi Engineering Research Center for Stem Cell, Shangrao, Jiangxi, China; Tianjin Key Laboratory of Engineering Technologies for Cell Pharmaceutical, National Engineering Research Center of Cell Products, AmCellGene Co., Ltd., Tianjin, China; Beijing Engineering Laboratory of Perinatal Stem Cells, Beijing Institute of Health and Stem Cells, Health & Biotech Co., Beijing, China
| | - Deling Kong
- State Key Laboratory of Medicinal Chemical Biology, The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, the College of Life Sciences, 94 Weijin Road, Tianjin 300071, China
| | - Zhikun Guo
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, 601 Jinsui Road, Xinxiang, Henan 453003, China.
| | - Qiang Zhao
- State Key Laboratory of Medicinal Chemical Biology, The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, the College of Life Sciences, 94 Weijin Road, Tianjin 300071, China.
| | - Zongjin Li
- Nankai University School of Medicine, 94 Weijin Road, Tianjin, China; State Key Laboratory of Medicinal Chemical Biology, The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, the College of Life Sciences, 94 Weijin Road, Tianjin 300071, China; State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, China; Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, 601 Jinsui Road, Xinxiang, Henan 453003, China.
| |
Collapse
|
5
|
Alizai PH, Bertram L, Kroy D, Kummer J, Andert A, Neumann UP, Ulmer TF, Fragoulis A. Expression of VEGFR-2 during Liver Regeneration after Partial Hepatectomy in a Bioluminescence Mouse Model. Eur Surg Res 2017; 58:330-340. [PMID: 29073598 DOI: 10.1159/000479628] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 07/19/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND Liver regeneration requires the formation of new blood vessels. Endothelial cell proliferation is stimulated by vascular endothelial growth factor (VEGF) and its receptor tyrosine kinase VEGFR-2. The aim of this study was to investigate VEGFR-2 expression in vivo during liver regeneration after partial hepatectomy (PHx). METHODS Transgenic VEGFR-2-luc mice were used in which the luciferase reporter gene was under control of the VEGFR-2 promoter. Following 2/3 PHx, the mice underwent in vivo bioluminescence imaging until the 14th postoperative day. Additionally, liver tissue was analyzed by immunohistochemistry, in vitro luminescence assays, and quantitative RT-PCR. RESULTS In vivo bioluminescence imaging showed a significant increase in VEGFR-2 promoter activity after PHx. Maximum signal was recorded on the 3rd day; 8 days postoperatively the signal intensity decreased significantly. On the 14th day, bioluminescence signal reached almost baseline levels. Immunohistochemistry, quantitative RT-PCR, and in vitro luminescence confirmed a significant increase on the 3rd day following resection. The mRNA expression of VEGFR-2 was significantly higher on day 3 than preoperatively as well as on day 8. CONCLUSION In vivo bioluminescence imaging with transgenic VEGFR-2-luc mice is feasible and provides a convenient model for noninvasively studying VEGFR-2 expression during liver regeneration. This may facilitate further experiments with modulation of angiogenesis by different substances.
Collapse
Affiliation(s)
- Patrick Hamid Alizai
- Department of General, Visceral and Transplantation Surgery, RWTH Aachen University Hospital, Aachen, Germany
| | - Lea Bertram
- Department of Surgery, Luisenhospital Aachen, Aachen, Germany.,Department of Anatomy and Cell Biology, RWTH Aachen University Hospital, Aachen, Germany
| | - Daniela Kroy
- Department of Gastroenterology and Metabolic Disorders, RWTH Aachen University Hospital, Aachen, Germany
| | - Julia Kummer
- Department of General, Visceral and Transplantation Surgery, RWTH Aachen University Hospital, Aachen, Germany.,Department of Gynaecology and Obstetrics, Vivantes Clinic Berlin, Berlin, Germany
| | - Anne Andert
- Department of General, Visceral and Transplantation Surgery, RWTH Aachen University Hospital, Aachen, Germany
| | - Ulf Peter Neumann
- Department of General, Visceral and Transplantation Surgery, RWTH Aachen University Hospital, Aachen, Germany
| | - Tom Florian Ulmer
- Department of General, Visceral and Transplantation Surgery, RWTH Aachen University Hospital, Aachen, Germany
| | - Athanassious Fragoulis
- Department of General, Visceral and Transplantation Surgery, RWTH Aachen University Hospital, Aachen, Germany.,Department of Anatomy and Cell Biology, RWTH Aachen University Hospital, Aachen, Germany
| |
Collapse
|
6
|
Huang J, Mei H, Tang Z, Li J, Zhang X, Lu Y, Huang F, Jin Q, Wang Z. Triple-amiRNA VEGFRs inhibition in pancreatic cancer improves the efficacy of chemotherapy through EMT regulation. J Control Release 2016; 245:1-14. [PMID: 27889393 DOI: 10.1016/j.jconrel.2016.11.024] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Accepted: 11/21/2016] [Indexed: 02/07/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a devastating disease with dismal outcome. Both novel prognostic markers and therapeutic targets are needed to improve the overall outcome of patients. Although single or double VEGFRs have been studied in PDAC, little is known about the role of triple combination of VEGFRs (VEGFR1, 2, and 3) in prognosis and therapy. We determined VEGFRs protein expression in 241 pancreatic tissues by tissue microarray immunohistochemistry (TMA-IHC), and correlated with patients' clinical characteristics and overall survival. Subsequently, we inactivated VEGFRs expression using artificial microRNAs (amiRNAs) in vitro. Triple combination of amiRNAs to VEGFRs reduced cell proliferation, increased apoptosis, and reduced cell migration and invasion in pancreatic cancer cell lines. In the mouse xenograft pancreatic cancer model, triple VEGFRs silencing significantly reduced tumor growth, had synergistic effect with standard chemotherapy, and was associated with inhibition of epithelial mesenchymal transition (EMT). We conclude that triple combination of VEGFRs is a prognostic marker for PDAC, and inhibition of VEGFRs expression via amiRNA represents a novel targeted therapy in PDAC through regulating EMT.
Collapse
Affiliation(s)
- Jianfei Huang
- Department of Clinical Bio-bank, Nantong University Affiliated Hospital, Nantong, Jiangsu 226001, China; Department of Pathology, Nantong University Affiliated Hospital, Nantong, Jiangsu 226001, China
| | - Haijun Mei
- Department of General Surgery, Nantong University Affiliated Hospital, Nantong, Jiangsu 226001, China
| | - Zhiyuan Tang
- Department of Respiratory Medicine Laboratory, Nantong University Affiliated Hospital, Nantong, Jiangsu 226001, China
| | - Jieying Li
- Department of Pathology, Nantong University Affiliated Hospital, Nantong, Jiangsu 226001, China
| | - Xiaojing Zhang
- Department of Pathology, Nantong University Affiliated Hospital, Nantong, Jiangsu 226001, China
| | - Yixiang Lu
- Maijie Biological Technology Co., Ltd. Nantong, Jiangsu 226007, China
| | - Fang Huang
- Department of Pathology, Nantong University Affiliated Hospital, Nantong, Jiangsu 226001, China
| | - Qin Jin
- Department of Pathology, Nantong University Affiliated Hospital, Nantong, Jiangsu 226001, China
| | - Zhiwei Wang
- Department of General Surgery, Nantong University Affiliated Hospital, Nantong, Jiangsu 226001, China.
| |
Collapse
|
7
|
Suero-Abreu GA, Aristizábal O, Bartelle BB, Volkova E, Rodríguez JJ, Turnbull DH. Multimodal Genetic Approach for Molecular Imaging of Vasculature in a Mouse Model of Melanoma. Mol Imaging Biol 2016; 19:203-214. [PMID: 27677887 DOI: 10.1007/s11307-016-1006-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
PURPOSE In this study, we evaluated a genetic approach for in vivo multimodal molecular imaging of vasculature in a mouse model of melanoma. PROCEDURES We used a novel transgenic mouse, Ts-Biotag, that genetically biotinylates vascular endothelial cells. After inoculating these mice with B16 melanoma cells, we selectively targeted endothelial cells with (strept)avidinated contrast agents to achieve multimodal contrast enhancement of Tie2-expressing blood vessels during tumor progression. RESULTS This genetic targeting system provided selective labeling of tumor vasculature and showed in vivo binding of avidinated probes with high specificity and sensitivity using microscopy, near infrared, ultrasound, and magnetic resonance imaging. We further demonstrated the feasibility of conducting longitudinal three-dimensional (3D) targeted imaging studies to dynamically assess changes in vascular Tie2 from early to advanced tumor stages. CONCLUSIONS Our results validated the Ts-Biotag mouse as a multimodal targeted imaging system with the potential to provide spatio-temporal information about dynamic changes in vasculature during tumor progression.
Collapse
Affiliation(s)
- Giselle A Suero-Abreu
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine (NYUSoM), 540 First Ave, New York, NY, 10016, USA
- Biomedical Imaging Graduate Program, NYUSoM, New York, NY, USA
- Department of Radiology, NYUSoM, New York, NY, USA
| | - Orlando Aristizábal
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine (NYUSoM), 540 First Ave, New York, NY, 10016, USA
| | - Benjamin B Bartelle
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine (NYUSoM), 540 First Ave, New York, NY, 10016, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Eugenia Volkova
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine (NYUSoM), 540 First Ave, New York, NY, 10016, USA
| | - Joe J Rodríguez
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine (NYUSoM), 540 First Ave, New York, NY, 10016, USA
| | - Daniel H Turnbull
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine (NYUSoM), 540 First Ave, New York, NY, 10016, USA.
- Biomedical Imaging Graduate Program, NYUSoM, New York, NY, USA.
- Department of Radiology, NYUSoM, New York, NY, USA.
- Department of Pathology, NYUSoM, New York, NY, USA.
| |
Collapse
|
8
|
O'Farrell AC, Shnyder SD, Marston G, Coletta PL, Gill JH. Non-invasive molecular imaging for preclinical cancer therapeutic development. Br J Pharmacol 2014; 169:719-35. [PMID: 23488622 DOI: 10.1111/bph.12155] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Revised: 01/02/2013] [Accepted: 02/10/2013] [Indexed: 12/18/2022] Open
Abstract
Molecular and non-invasive imaging are rapidly emerging fields in preclinical cancer drug discovery. This is driven by the need to develop more efficacious and safer treatments, the advent of molecular-targeted therapeutics, and the requirements to reduce and refine current preclinical in vivo models. Such bioimaging strategies include MRI, PET, single positron emission computed tomography, ultrasound, and optical approaches such as bioluminescence and fluorescence imaging. These molecular imaging modalities have several advantages over traditional screening methods, not least the ability to quantitatively monitor pharmacodynamic changes at the cellular and molecular level in living animals non-invasively in real time. This review aims to provide an overview of non-invasive molecular imaging techniques, highlighting the strengths, limitations and versatility of these approaches in preclinical cancer drug discovery and development.
Collapse
Affiliation(s)
- A C O'Farrell
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | | | | | | | | |
Collapse
|
9
|
Levin RA, Felsen CN, Yang J, Lin JY, Whitney MA, Nguyen QT, Tsien RY. An optimized triple modality reporter for quantitative in vivo tumor imaging and therapy evaluation. PLoS One 2014; 9:e97415. [PMID: 24816650 PMCID: PMC4016317 DOI: 10.1371/journal.pone.0097415] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 04/17/2014] [Indexed: 11/24/2022] Open
Abstract
We present an optimized triple modality reporter construct combining a far-red fluorescent protein (E2-Crimson), enhanced firefly luciferase enzyme (Luc2), and truncated wild type herpes simplex virus I thymidine kinase (wttk) that allows for sensitive, long-term tracking of tumor growth in vivo by fluorescence, bioluminescence, and positron emission tomography. Two human cancer cell lines (MDA-MB-231 breast cancer and HT-1080 fibrosarcoma cancer) were successfully transduced to express this triple modality reporter. Fluorescence and bioluminescence imaging of the triple modality reporter were used to accurately quantify the therapeutic responses of MDA-MB-231 tumors to the chemotherapeutic agent monomethyl auristatin E in vivo in athymic nude mice. Positive correlation was observed between the fluorescence and bioluminescence signals, and these signals were also positively correlated with the ex vivo tumor weights. This is the first reported use of both fluorescence and bioluminescence signals from a multi-modality reporter construct to measure drug efficacy in vivo.
Collapse
Affiliation(s)
- Rachel A. Levin
- Department of Pharmacology, UCSD School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Csilla N. Felsen
- Department of Pharmacology, UCSD School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Jin Yang
- Department of Pharmacology, UCSD School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - John Y. Lin
- Department of Pharmacology, UCSD School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Michael A. Whitney
- Department of Pharmacology, UCSD School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Quyen T. Nguyen
- Division of Otolaryngology/Head and Neck Surgery, University of California San Diego, La Jolla, California, United States of America
| | - Roger Y. Tsien
- Department of Pharmacology, UCSD School of Medicine, University of California San Diego, La Jolla, California, United States of America
- Howard Hughes Medical Institute, La Jolla, California, United States of America
| |
Collapse
|
10
|
Zhao D, Tu Y, Wan L, Bu L, Huang T, Sun X, Wang K, Shen B. In vivo monitoring of angiogenesis inhibition via down-regulation of mir-21 in a VEGFR2-luc murine breast cancer model using bioluminescent imaging. PLoS One 2013; 8:e71472. [PMID: 23951172 PMCID: PMC3738509 DOI: 10.1371/journal.pone.0071472] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 06/29/2013] [Indexed: 01/04/2023] Open
Abstract
MicroRNA-21 (miR-21) is overexpressed in a wide range of cancers and involved in tumor proliferation and metastasis. However, the potential function of miR-21 in regulating tumor angiogenesis has been little disclosed. In this study, we treated the cultured 4T1 murine breast cancer cells and human umbilical vein endothelial cells (HUVECs) with miR-21 mimic, antagomir-21 or negative control (scramble), which were subjected to MTT, terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL), quantitative Reverse Transcriptase PCR (qRT-PCR) and immunoblotting analysis. In addition, 4T1 cells were implanted beneath the right breast fat pad of the VEGFR2-luc transgenic mice, which were randomly divided into three groups and received saline, antagomir-21 or scramble treatment once respectively after tumor model establishment. Bioluminescent imaging was used to monitor tumor growth and angiogenesis in vivo at 0d, 3d, 5d, 7d, 10d, and 14d after treatment. Mice were killed at the end of study and tumor tissues were collected for use. The results showed that knockdown of miR-21 by antagomir-21 decreased cell proliferation and induced apoptosis via targeting PTEN both in 4T1 cells and HUVECs. We also found the anti-angiogenesis and anti-tumor effects of antagomir-21 in the VEGFR2-luc transgenic mouse model using bioluminescent imaging. Moreover, the Western blotting data revealed that antagomir-21 inhibited tumor angiogenesis through suppressing HIF-1α/VEGF/VEGFR2-associated signaling pathway. In conclusion, the results from current study demonstrate that antagomir-21 can effectively suppress tumor growth and angiogenesis in VEGFR2-luc mouse breast tumor model and bioluminescent imaging can be used as a tool for noninvasively and continuously monitoring tumor angiogenesis in vivo.
Collapse
Affiliation(s)
- Dongliang Zhao
- Department of Radiology, the Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Key Laboratory of Molecular Imaging, College of Heilongjiang Province, Harbin, Heilongjiang, China
| | - Yingfeng Tu
- Department of Radiology, the Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Key Laboratory of Molecular Imaging, College of Heilongjiang Province, Harbin, Heilongjiang, China
- Department of Cardiology, the Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Lin Wan
- Department of Radiology, the Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Key Laboratory of Molecular Imaging, College of Heilongjiang Province, Harbin, Heilongjiang, China
| | - Lihong Bu
- Department of Radiology, the Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Key Laboratory of Molecular Imaging, College of Heilongjiang Province, Harbin, Heilongjiang, China
| | - Tao Huang
- Department of Radiology, the Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Key Laboratory of Molecular Imaging, College of Heilongjiang Province, Harbin, Heilongjiang, China
| | - Xilin Sun
- Department of Radiology, the Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Key Laboratory of Molecular Imaging, College of Heilongjiang Province, Harbin, Heilongjiang, China
| | - Kai Wang
- Department of Radiology, the Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Key Laboratory of Molecular Imaging, College of Heilongjiang Province, Harbin, Heilongjiang, China
| | - Baozhong Shen
- Department of Radiology, the Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Key Laboratory of Molecular Imaging, College of Heilongjiang Province, Harbin, Heilongjiang, China
- * E-mail:
| |
Collapse
|
11
|
Chai MG, Kim-Fuchs C, Angst E, Sloan EK. Bioluminescent orthotopic model of pancreatic cancer progression. J Vis Exp 2013. [PMID: 23852391 DOI: 10.3791/50395] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Pancreatic cancer has an extremely poor five-year survival rate of 4-6%. New therapeutic options are critically needed and depend on improved understanding of pancreatic cancer biology. To better understand the interaction of cancer cells with the pancreatic microenvironment, we demonstrate an orthotopic model of pancreatic cancer that permits non-invasive monitoring of cancer progression. Luciferase-tagged pancreatic cancer cells are resuspended in Matrigel and delivered into the pancreatic tail during laparotomy. Matrigel solidifies at body temperature to prevent leakage of cancer cells during injection. Primary tumor growth and metastasis to distant organs are monitored following injection of the luciferase substrate luciferin, using in vivo imaging of bioluminescence emission from the cancer cells. In vivo imaging also may be used to track primary tumor recurrence after resection. This orthotopic model is suited to both syngeneic and xenograft models and may be used in pre-clinical trials to investigate the impact of novel anti-cancer therapeutics on the growth of the primary pancreatic tumor and metastasis.
Collapse
Affiliation(s)
- Ming G Chai
- Monash Institute of Pharmaceutical Sciences, Monash University
| | | | | | | |
Collapse
|
12
|
Wang W, Liu H, Song Z, Wang Y, Wang J, Li Y, Li Y, Wang M, Chen J, Zhou Q. [Identification of the offspring of Vegfr2-luc transgenic mouse]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2011; 14:391-5. [PMID: 21569642 PMCID: PMC6000322 DOI: 10.3779/j.issn.1009-3419.2011.05.02] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND AND OBJECTIVE A transgenic mouse, Vegfr2-luc, in which a luciferase reporter (luc) is under control of the murine VEGFR2 promoter, can be used to track angiogenesis in vivo. The aim of this study is to identify the offspring of Vegfr2-luc transgenic mouse. METHODS Luc was detected with PCR in genomic DNA of the new-born mouse. Luc expression in the offspring of Vegfr2-luc transgenic mouse was monitored with IVIS in vivo imaging system during post-natal development. Wound-healing models of Vegfr2-luc transgenic mouse offspring were established and the expression of luc was monitored during the wound-healing process. Luc activity and VEGFR2 mRNA expression in different organs were detected with luc Assay System and Real-time PCR respectively. RESULTS PCR showed that 50% (56/112) of the offspring of Vegfr2-luc transgenic mouse carry luc. IVIS in vivo imaging results demonstrated that luc expression in Vegfr2-luc transgenic mouse dropped dramatically with age increase (P<0.001) and luc expression in the wound first increased and then decreased during the wound-healing process (P<0.001). Luc activity in female Vegfr2-luc transgenic mouse organs was positively correlated with VEGFR2 mRNA expression (r=0.948, P<0.001). Except testis, luc activity in male Vegfr2-luc transgenic mouse organs was also positively correlated with VEGFR2 mRNA expression (r=0.836, P<0.001). CONCLUSION The offspring of Vegfr2-luc transgenic mouse is applicable to tracking angiogenesis in vivo.
Collapse
Affiliation(s)
- Weiqiang Wang
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenviroment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Greene JM, Dunaway CW, Bowers SD, Rude BJ, Feugang JM, Ryan PL. In vivo monitoring of fetoplacental Vegfr2 gene activity in a murine pregnancy model using a Vegfr2-luc reporter gene and bioluminescent imaging. Reprod Biol Endocrinol 2011; 9:51. [PMID: 21496302 PMCID: PMC3084164 DOI: 10.1186/1477-7827-9-51] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2010] [Accepted: 04/16/2011] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Vascular endothelial growth factor receptor-2 (VEGFR2) plays a pivotal role in angiogenesis by eliciting vascular endothelial cell growth when bound to VEGF, a powerful pro-angiogenic ligand. While Vegf and Vegfr2 are expressed throughout gestation, the latter third of gestation in mice is characterized by a marked increase in fetoplacental angiogenesis. Thus, the objective of this study was to determine the feasibility of monitoring fetoplacental Vegfr2 gene activity non-invasively using a Vegfr2-luc reporter transgenic mouse and bioluminescent imaging. METHODS Imaging parameters were optimized using two wild-type (WT) females, bearing Vegfr2-luc fetuses. Then, seven WT females, bred to Vegfr2-luc males, were imaged from gestational day (GD) 12 to 18 to determine the usefulness of the Vegfr2-luc mouse as a model for studying fetoplacental Vegfr2 activity during pregnancy. Semi-quantitative RT-PCR of Vegfr2 was also performed on whole fetoplacental units during this time. Additionally, resultant neonates were imaged at postnatal day (PND) 7, 14 and 21 to monitor Vegfr2 activity during post-natal development. RESULTS Fetoplacental Vegfr2 gene activity was detected as light emissions beginning on GD 12 of gestation and increased throughout the imaging period (P < 0.05), and this paralleled the Vegfr2 mRNA data obtained from RT-PCR analysis. A decline in fetoplacental light emissions was associated with a poor pregnancy outcome in one pregnancy, indicating that this approach has potential use for studies monitoring pregnancy well being. Additionally, neonatal Vegfr2 activity was detected at PND 7, 14 and 21 but declined with time (P < 0.0001). CONCLUSIONS In utero fetoplacental Vegfr2 gene activity was monitored longitudinally in a quantitative manner using a luciferase reporter gene and bioluminescent imaging during the latter third of gestation. This study demonstrates the feasibility of using the Vegfr2-luc mouse to monitor late gestation fetoplacental angiogenic activity under normal and experimental conditions. Additionally, neonatal Vegfr2 gene activity was monitored for three weeks postpartum, allowing continuous monitoring of Vegfr2 activity during the latter third of gestation and postnatal development within the same animals.
Collapse
Affiliation(s)
- Jonathan M Greene
- Department of Pathobiology and Population Medicine, Mississippi State University College of Veterinary Medicine, Mississippi State, MS, USA
- Facility for Organismal and Cellular Imaging, Mississippi State University, Mississippi State, MS, USA
| | - Chad W Dunaway
- Facility for Organismal and Cellular Imaging, Mississippi State University, Mississippi State, MS, USA
| | - Susan D Bowers
- Facility for Organismal and Cellular Imaging, Mississippi State University, Mississippi State, MS, USA
- Department of Animal and Dairy Sciences, Mississippi State University, Mississippi State, MS, USA
| | - Brian J Rude
- Department of Animal and Dairy Sciences, Mississippi State University, Mississippi State, MS, USA
| | - Jean M Feugang
- Facility for Organismal and Cellular Imaging, Mississippi State University, Mississippi State, MS, USA
- Department of Animal and Dairy Sciences, Mississippi State University, Mississippi State, MS, USA
| | - Peter L Ryan
- Department of Pathobiology and Population Medicine, Mississippi State University College of Veterinary Medicine, Mississippi State, MS, USA
- Facility for Organismal and Cellular Imaging, Mississippi State University, Mississippi State, MS, USA
- Department of Animal and Dairy Sciences, Mississippi State University, Mississippi State, MS, USA
| |
Collapse
|
14
|
Angst E, Dawson DW, Stroka D, Gloor B, Park J, Candinas D, Reber HA, Hines OJ, Eibl G. N-myc downstream regulated gene-1 expression correlates with reduced pancreatic cancer growth and increased apoptosis in vitro and in vivo. Surgery 2011; 149:614-24. [PMID: 21236457 DOI: 10.1016/j.surg.2010.11.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2010] [Accepted: 11/09/2010] [Indexed: 01/22/2023]
Abstract
BACKGROUND The role of N-myc downstream regulated gene-1 (NDRG1) in cancer has recently gained interest, as potential regulator of cell death and tumor suppressor. Although its normal function in the pancreas is largely unknown, loss of NDRG1 expression is associated with a more aggressive tumor phenotype and poor outcome in pancreatic cancer patients. METHODS NDRG1 expression was determined in human pancreatic cancer samples and across a panel of 6 pancreatic cancer cell lines. HPAF-II cells were stably transfected with full-length NDRG1 (HP(NDRG1)) or the empty vector (HP(NEG)). BxPC-3 cells were silenced for NDRG1. These cells were analyzed for cell growth, cell cycle, apoptosis, and pro-apoptotic gene expression. They were transduced with lentiviral vector expressing luciferase to establish an orthotopic xenograft model. In vivo tumor growth was assessed by bioluminescence imaging. RESULTS Growth of HP(NDRG1) was impaired in anchorage-dependent and abolished in anchorage-independent assays respectively. These results were confirmed in BxPC-3 silenced for NDRG1. Growth inhibition was induced by enhanced apoptosis. Seven pro-apoptotic genes were up-regulated in HP(NDRG1) cells. In vivo, HP(NDRG1) tumors showed no growth over 6 weeks, while HP(NEG) tumors grew prominently, correlating with a 325-fold increase in light emission. CONCLUSION In this model we found that expression of NDRG1 correlates with decreased growth in human pancreatic cancer cells in vitro and in vivo. The observation that NDRG1 completely inhibited growth in anchorage-independent assays and in vivo supports a biological function as a tumor suppressor in pancreatic cancer.
Collapse
Affiliation(s)
- Eliane Angst
- Hirshberg Laboratories for Pancreatic Cancer Research, Department of Surgery, UCLA Center for Excellence in Pancreatic Diseases, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | | | | | | | | | | | | | | | | |
Collapse
|